1
|
Hestehave S, Allen HN, Gomez K, Duran P, Calderon-Rivera A, Loya-López S, Rodríguez-Palma EJ, Khanna R. Small molecule targeting Na V 1.7 via inhibition of CRMP2-Ubc9 interaction reduces pain-related outcomes in a rodent osteoarthritic model. Pain 2025; 166:99-111. [PMID: 39106443 DOI: 10.1097/j.pain.0000000000003357] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/11/2024] [Accepted: 05/30/2024] [Indexed: 08/09/2024]
Abstract
ABSTRACT Osteoarthritis (OA) is a highly prevalent and disabling joint disease, characterized by pathological progressive joint deformation and clinical symptoms of pain. Disease-modifying treatments remain unavailable, and pain-mitigation is often suboptimal, but recent studies suggest beneficial effects by inhibition of the voltage-gated sodium channel Na V 1.7. We previously identified compound 194 as an indirect inhibitor of Na V 1.7 by preventing SUMOylation of the Na V 1.7-trafficking protein, collapsin response mediator protein 2. Compound 194 reduces the functional activity of Na V 1.7 channels and produces effective analgesia in a variety of acute and neuropathic pain models. However, its effectiveness has not yet been evaluated in models of OA. Here, we explore the effects of 194 on pain-related outcomes in the OA-like monoiodoacetate model using behavioral assessment, biochemistry, novel in vivo fiber photometry, and patch clamp electrophysiology. We found that the monoiodoacetate model induced (1) increased pain-like behaviors and calcium responses of glutamatergic neurons in the parabrachial nucleus after evoked cold and mechanical stimuli, (2) conditioned place aversion to mechanical stimulation, (3) functional weight bearing asymmetry, (4) increased sodium currents in dorsal root ganglia neurons, and (5) increased calcitonin gene-related peptide-release in the spinal cord. Crucially, administration of 194 improved all these pain-related outcomes. Collectively, these findings support indirect inhibition of Na V 1.7 as an effective treatment of OA-related pain through the inhibition of collapsin response mediator protein 2-SUMOylation via compound 194.
Collapse
Affiliation(s)
- Sara Hestehave
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, United States
- Pain Research Center, New York University, New York, NY, United States . Dr. Hestehave is now with the Department of Experimental Medicine, University of Copenhagen, Copenhagen N, Denmark. Dr. Allen, Dr. Gomez, Dr. Calderon-Rivera, Dr. Loya-López, Dr. Rodríguez-Palma, and Dr. Khanna are now with the Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Heather N Allen
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, United States
- Pain Research Center, New York University, New York, NY, United States . Dr. Hestehave is now with the Department of Experimental Medicine, University of Copenhagen, Copenhagen N, Denmark. Dr. Allen, Dr. Gomez, Dr. Calderon-Rivera, Dr. Loya-López, Dr. Rodríguez-Palma, and Dr. Khanna are now with the Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Kimberly Gomez
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, United States
- Pain Research Center, New York University, New York, NY, United States . Dr. Hestehave is now with the Department of Experimental Medicine, University of Copenhagen, Copenhagen N, Denmark. Dr. Allen, Dr. Gomez, Dr. Calderon-Rivera, Dr. Loya-López, Dr. Rodríguez-Palma, and Dr. Khanna are now with the Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Paz Duran
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, United States
- Pain Research Center, New York University, New York, NY, United States . Dr. Hestehave is now with the Department of Experimental Medicine, University of Copenhagen, Copenhagen N, Denmark. Dr. Allen, Dr. Gomez, Dr. Calderon-Rivera, Dr. Loya-López, Dr. Rodríguez-Palma, and Dr. Khanna are now with the Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Aida Calderon-Rivera
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, United States
- Pain Research Center, New York University, New York, NY, United States . Dr. Hestehave is now with the Department of Experimental Medicine, University of Copenhagen, Copenhagen N, Denmark. Dr. Allen, Dr. Gomez, Dr. Calderon-Rivera, Dr. Loya-López, Dr. Rodríguez-Palma, and Dr. Khanna are now with the Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Santiago Loya-López
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, United States
- Pain Research Center, New York University, New York, NY, United States . Dr. Hestehave is now with the Department of Experimental Medicine, University of Copenhagen, Copenhagen N, Denmark. Dr. Allen, Dr. Gomez, Dr. Calderon-Rivera, Dr. Loya-López, Dr. Rodríguez-Palma, and Dr. Khanna are now with the Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Erick J Rodríguez-Palma
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, United States
- Pain Research Center, New York University, New York, NY, United States . Dr. Hestehave is now with the Department of Experimental Medicine, University of Copenhagen, Copenhagen N, Denmark. Dr. Allen, Dr. Gomez, Dr. Calderon-Rivera, Dr. Loya-López, Dr. Rodríguez-Palma, and Dr. Khanna are now with the Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| | - Rajesh Khanna
- Department of Molecular Pathobiology, College of Dentistry, New York University, New York, NY, United States
- Pain Research Center, New York University, New York, NY, United States . Dr. Hestehave is now with the Department of Experimental Medicine, University of Copenhagen, Copenhagen N, Denmark. Dr. Allen, Dr. Gomez, Dr. Calderon-Rivera, Dr. Loya-López, Dr. Rodríguez-Palma, and Dr. Khanna are now with the Department of Pharmacology & Therapeutics, University of Florida College of Medicine, Gainesville, FL, United States
| |
Collapse
|
2
|
Kauppinen S, Fercher D, Barreto G, Karjalainen VP, Virtanen V, Baixauli-Marin L, Fonti M, Zhang S, Frondelius T, Weber P, Saarakkala S, Zenobi-Wong M, Finnilä MAJ. Assessment of whole cartilage surface damage in an osteoarthritis rat model: The Cartilage Roughness Score (CRS) utilizing microcomputed tomography. Osteoarthritis Cartilage 2025; 33:134-145. [PMID: 39357597 DOI: 10.1016/j.joca.2024.09.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 01/25/2024] [Revised: 08/27/2024] [Accepted: 09/23/2024] [Indexed: 10/04/2024]
Abstract
OBJECTIVE This study aims to establish an accurate and robust imaging biomarker for pre-clinical osteoarthritis (OA) research, focusing on early detection of cartilage surface degeneration. METHOD Using 50 male Wistar rats, this study aims to observe Collagenase-induced OA (CIOA) progression through microcomputed x-ray tomography (µCT), histopathological analysis, and gait analysis. A novel parameter, Cartilage Roughness Score (CRS), was developed for assessing cartilage structural damage from µCT data and was compared with histological OARSI Cartilage Degeneration Score (OARSI CDS). Additionally, as CRS maps the full surface, it was used to simulate the level of uncertainty in histological sampling. RESULTS CRS and OARSI CDS have a linear relationship. CRS for healthy cartilage is 2.75 (95% CI: 1.14-4.36), and with every 1 unit increase in OARSI, CRS is expected to increase by 0.64 (95% CI: 0.35-0.92). Cartilage degeneration due to CIOA was evident in both histopathological scoring and CRS. However, only CRS was sensitive enough to show consistent damage progression from day 10 to day 60. Furthermore, our simulation for histological sampling suggested that up to 16 coronal slices with 200 µm spacing would be needed to accurately represent the full extent of cartilage surface degeneration in a slice-wise manner. Gait analysis showed changes solely at eight days post-collagenase injection, normalizing by day 60. CONCLUSION The CRS analysis method emerges as a robust tool for cartilage surface damage assessment. This study demonstrates the potential of automatic 3D analysis over the traditional 2D histological approach when evaluating cartilage surface damage.
Collapse
Affiliation(s)
- Sami Kauppinen
- Research unit of Health Sciences and Technology, University of Oulu, Finland.
| | - David Fercher
- Tissue Engineering and Biofabrication, ETH Zürich, Switzerland
| | - Gonçalo Barreto
- Clinicum, Faculty of Medicine, University of Helsinki and Helsinki University Hospital, Helsinki, Finland
| | | | - Vesa Virtanen
- Research unit of Health Sciences and Technology, University of Oulu, Finland
| | | | - Marina Fonti
- Tissue Engineering and Biofabrication, ETH Zürich, Switzerland
| | - Shipin Zhang
- Tissue Engineering and Biofabrication, ETH Zürich, Switzerland
| | - Tuomas Frondelius
- Research unit of Health Sciences and Technology, University of Oulu, Finland
| | - Patrick Weber
- Tissue Engineering and Biofabrication, ETH Zürich, Switzerland
| | - Simo Saarakkala
- Research unit of Health Sciences and Technology, University of Oulu, Finland
| | | | - Mikko A J Finnilä
- Research unit of Health Sciences and Technology, University of Oulu, Finland
| |
Collapse
|
3
|
Horie T, Hirata H, Sakamoto T, Kitajima H, Fuku A, Nakamura Y, Sunatani Y, Tanida I, Sunami H, Tachi Y, Ishigaki Y, Yamamoto N, Shimizu Y, Ichiseki T, Kaneuji A, Iwabuchi K, Osawa S, Kawahara N. Multiomics analyses reveal adipose-derived stem cells inhibit the inflammatory response of M1-like macrophages through secreting lactate. Stem Cell Res Ther 2024; 15:485. [PMID: 39696485 DOI: 10.1186/s13287-024-04072-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2023] [Accepted: 11/20/2024] [Indexed: 12/20/2024] Open
Abstract
BACKGROUND Adipose-derived stem cells (ADSCs) are widely used in the field of regenerative medicine because of their various functions, including anti-inflammatory effects. ADSCs are considered to exert their anti-inflammatory effects by secreting anti-inflammatory cytokines and extracellular vesicles. Although recent studies have reported that metabolites have a variety of physiological activities, whether those secreted by ADSCs have anti-inflammatory properties remains unclear. Here, we performed multiomics analyses to examine the effect of ADSC-derived metabolites on M1-like macrophages, which play an important role in inflammatory responses. METHODS The concentration of metabolites in the culture supernatant of ADSCs was quantified using capillary electrophoresis time-of-flight mass spectrometry. To evaluate their effects on inflammatory responses, M1-like macrophages were exposed to the conditioned ADSC medium or their metabolites, and RNA sequencing was used to detect gene expression changes. Immunoblotting was performed to examine how the metabolite suppresses inflammatory processes. To clarify the contribution of the metabolite in the conditioned medium to its anti-inflammatory effects, metabolite uptake was pharmacologically inhibited, and gene expression and the tumor necrosis factor-α concentration were measured by quantitative PCR and enzyme-linked immunosorbent assay, respectively. RESULTS Metabolomic analysis showed large amounts of lactate in the culture supernatant. The conditioned medium and lactate significantly suppressed or increased the pro-inflammatory and anti-inflammatory gene expressions. However, sequencing and immunoblotting analysis revealed that lactate did not induce polarization from M1- to M2-like macrophages. Based on a recent report that the immunosuppressive effect of lactate depends on epigenetic reprogramming, histone acetylation was investigated, and H3K27ac expression was upregulated. In addition, 7ACC2, which specifically inhibits the monocarboxylate transporter 1, significantly inhibited the anti-inflammatory effect of the conditioned ADSC medium on M1-like macrophages. CONCLUSIONS Our results showed that ADSCs suppress pro-inflammatory effects of M1-like macrophages by secreting lactate. This study adds to our understanding of the importance of metabolites and is also expected to elucidate new mechanisms of ADSC treatments.
Collapse
Affiliation(s)
- Tetsuhiro Horie
- Medical Research Institute, Kanazawa Medical University, Kahoku, Ishikawa, 920-0293, Japan
- Department of Pharmacy, Kanazawa Medical University Hospital, Kahoku, Ishikawa, 920-0293, Japan
| | - Hiroaki Hirata
- Department of Orthopedic Surgery, Kanazawa Medical University, Kahoku, Ishikawa, 920-0293, Japan.
| | - Takuya Sakamoto
- Medical Research Institute, Kanazawa Medical University, Kahoku, Ishikawa, 920-0293, Japan
- Department of Pharmacy, Kanazawa Medical University Hospital, Kahoku, Ishikawa, 920-0293, Japan
| | - Hironori Kitajima
- Department of Orthopedic Surgery, Kanazawa Medical University, Kahoku, Ishikawa, 920-0293, Japan
| | - Atsushi Fuku
- Department of Orthopedic Surgery, Kanazawa Medical University, Kahoku, Ishikawa, 920-0293, Japan
| | - Yuka Nakamura
- Medical Research Institute, Kanazawa Medical University, Kahoku, Ishikawa, 920-0293, Japan
| | - Yumi Sunatani
- Department of Biochemistry I, Kanazawa Medical University, Kahoku, Ishikawa, 920-0293, Japan
| | - Ikuhiro Tanida
- Genome Biotechnology Laboratory, Kanazawa Institute of Technology, Hakusan, Ishikawa, 924-0838, Japan
| | - Hiroshi Sunami
- Advanced Medical Research Center, Faculty of Medicine, University of the Ryukyus, Nakagami, Okinawa, 903-0215, Japan
| | - Yoshiyuki Tachi
- Department of Orthopedic Surgery, Kanazawa Medical University, Kahoku, Ishikawa, 920-0293, Japan
| | - Yasuhito Ishigaki
- Medical Research Institute, Kanazawa Medical University, Kahoku, Ishikawa, 920-0293, Japan
| | - Naoki Yamamoto
- Research Promotion Headquarters, Fujita Health University, Toyoake, Aichi, 470-1192, Japan
| | - Yusuke Shimizu
- Department of Plastic and Reconstructive Surgery, Graduate School of Medicine, University of the Ryukyus, Nakagami, Okinawa, 903-0215, Japan
| | - Toru Ichiseki
- Department of Orthopedic Surgery, Kanazawa Medical University, Kahoku, Ishikawa, 920-0293, Japan.
| | - Ayumi Kaneuji
- Department of Orthopedic Surgery, Kanazawa Medical University, Kahoku, Ishikawa, 920-0293, Japan
| | - Kuniyoshi Iwabuchi
- Department of Biochemistry I, Kanazawa Medical University, Kahoku, Ishikawa, 920-0293, Japan
| | - Satoshi Osawa
- Genome Biotechnology Laboratory, Kanazawa Institute of Technology, Hakusan, Ishikawa, 924-0838, Japan
| | - Norio Kawahara
- Department of Orthopedic Surgery, Kanazawa Medical University, Kahoku, Ishikawa, 920-0293, Japan
| |
Collapse
|
4
|
Scanzello CR, Hasty KA, Chung CB, Griffin TM, Willet NJ, Krug H, Chu CQ, Ewart D, Jerban S, Baker JF, Duvall CL, Brunger JM, Burdick JA, Spindler KP, Drissi H. Teaming up to overcome challenges toward translation of new therapeutics for osteoarthritis. J Orthop Res 2024; 42:2659-2672. [PMID: 39103981 DOI: 10.1002/jor.25944] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 04/02/2024] [Revised: 06/18/2024] [Accepted: 07/09/2024] [Indexed: 08/07/2024]
Abstract
As a leading global cause of musculoskeletal-related disability, osteoarthritis (OA) represents a public health urgency. Understanding of disease pathogenesis has advanced substantially in the past decade, yet no disease-modifying therapeutics have advanced to the clinic. To address this challenge, the CARE-AP (Cartilage Repair strategies to alleviate Arthritis Pain) collaborative research team was convened to bring together relevant multidisciplinary expertise and perspectives from across the VA research community nationwide. The first CARE-AP Annual Research Symposium took place (virtually) in February 2022 with roughly 90 participants. A number of innovative and therapeutic strategies were discussed, including siRNA approaches coupled with novel nanoparticle-based delivery systems, cellular engineering approaches to develop reparative cells that can probe the joint environment and respond to disease-specific cues, and novel biofabrication techniques to improve tissue engineering and effect "biological joint replacement." In addition, challenges and advances in rehabilitation approaches, imaging outcomes, and clinical studies were presented, which were integrated into a framework of recommendations for running "preclinical trials" to improve successful clinical translation.
Collapse
Affiliation(s)
- Carla R Scanzello
- Translational Musculoskeletal Research Center, Corp. Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania, USA
- Division of Rheumatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Karen A Hasty
- Research Service 151, Lt. Col. Luke Weathers, Jr. VA Medical Center, Memphis, Tennessee, USA
- Department of Orthopaedic Surgery and Biomedical Engineering, Campbell Clinic/University of Tennessee Health Science Center, Memphis, Tennessee, USA
| | - Christine B Chung
- Radiology Service, Veterans Affairs San Diego Healthcare System, San Diego, California, USA
- Department of Radiology, University of California San Diego, La Jolla, California, USA
| | - Timothy M Griffin
- Oklahoma City VA Health Care System, Oklahoma City, Oklahoma, USA
- Aging and Metabolism Research Program, Oklahoma Medical Research Foundation, Oklahoma City, Oklahoma, USA
| | - Nick J Willet
- Veterans Affairs Portland Health Care System, Portland, Oregon, USA
- Phil and Penny Knight Campus for Accelerating Scientific Impact, Department of Bioengineering, University of Oregon, Eugene, Oregon, USA
| | - Hollis Krug
- Rheumatology Section, Minneapolis Veterans Affairs Medical Center, Minneapolis, Minnesota, USA
- Division of Rheumatology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Cong-Qiu Chu
- Veterans Affairs Portland Health Care System, Portland, Oregon, USA
- Division of Arthritis and Rheumatic Diseases, Oregon Health Sciences University, Portland, Oregon, USA
| | - David Ewart
- Rheumatology Section, Minneapolis Veterans Affairs Medical Center, Minneapolis, Minnesota, USA
- Division of Rheumatology, University of Minnesota, Minneapolis, Minnesota, USA
| | - Saeed Jerban
- Radiology Service, Veterans Affairs San Diego Healthcare System, San Diego, California, USA
- Department of Radiology, University of California San Diego, La Jolla, California, USA
| | - Joshua F Baker
- Translational Musculoskeletal Research Center, Corp. Michael J. Crescenz VA Medical Center, Philadelphia, Pennsylvania, USA
- Division of Rheumatology, University of Pennsylvania Perelman School of Medicine, Philadelphia, Pennsylvania, USA
| | - Craig L Duvall
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
| | - Jonathan M Brunger
- Department of Biomedical Engineering, Vanderbilt University, Nashville, Tennessee, USA
- Center for Stem Cell Biology, Vanderbilt University, Nashville, Tennessee, USA
| | - Jason A Burdick
- BioFrontiers Institute and Department of Chemical and Biological Engineering, University of Colorado, Boulder, Colorado, USA
| | - Kurt P Spindler
- Department of Orthopaedic Surgery, Sports Medicine, Cleveland Clinic Florida, Coral Springs, Florida, USA
| | - Hicham Drissi
- Department of Orthopaedics, Emory University School of Medicine, Atlanta, Georgia, USA
- Atlanta VA Medical Center, Decatur, Georgia, USA
| |
Collapse
|
5
|
Sasaki Y, Kijima K, Yoshioka K. Validity evaluation of a rat model of monoiodoacetate-induced osteoarthritis with clinically effective drugs. BMC Musculoskelet Disord 2024; 25:975. [PMID: 39609755 PMCID: PMC11605887 DOI: 10.1186/s12891-024-08083-9] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 07/24/2024] [Accepted: 11/15/2024] [Indexed: 11/30/2024] Open
Abstract
BACKGROUND Knee osteoarthritis (KOA) is the most common type of joint disease in elderly people and is characterized by pain and dysfunction. Although the monoiodoacetate (MIA)-induced model is widely used as a rodent KOA model, it is important to acknowledge the inherent limitations of this model, as the MIA model develops complex pathological phases on a daily basis. An accurate understanding of this model and the selection of an appropriate time point according to the target for drug candidates can lead to the development of clinically effective drugs. METHODS Changes in the pathological state of the MIA model were assessed via histopathological evaluation. Clodronate, a bisphosphonate, and diclofenac, a nonsteroidal anti-inflammatory drug (NSAID), were selected as models of clinically effective drugs due to their different mechanisms of action. The analgesic effects of both drugs on the MIA model were evaluated. The long-term effect of clodronate on subchondral bone osteoclasts was also evaluated. RESULTS Histopathological evaluation revealed that MIA-induced symptomatic behavior occurred in the early and late phases and was accompanied by synovial inflammation and osteoclast-related joint degeneration, respectively. Although clodronate inhibited symptomatic behavior and prevented cartilage degeneration from the early to late phases, diclofenac inhibited symptomatic behavior only in the early phase. Clodronate acted locally and inhibited the activation of subchondral osteoclasts. CONCLUSIONS Pathological changes, such as synovial changes in the early phase and knee joint degeneration in the late phase, in the MIA model are similar to those in human KOA. Our results indicate that the early phase in the MIA model is appropriate for evaluating the effects of anti-inflammatory agents such as NSAIDs and corticosteroids. The late phase in the MIA model is appropriate for evaluating the effects of drugs that act on cartilage and subchondral bone.
Collapse
Affiliation(s)
- Yamato Sasaki
- Central Research Laboratory, Research & Development Division, Seikagaku Corporation, Tateno 3-1253, Higashiyamato-shi, Tokyo, 207-0021, Japan.
| | - Kei Kijima
- Central Research Laboratory, Research & Development Division, Seikagaku Corporation, Tateno 3-1253, Higashiyamato-shi, Tokyo, 207-0021, Japan
| | - Keiji Yoshioka
- Central Research Laboratory, Research & Development Division, Seikagaku Corporation, Tateno 3-1253, Higashiyamato-shi, Tokyo, 207-0021, Japan
| |
Collapse
|
6
|
Moon J, Cho KH, Jhun J, Choi J, Na HS, Lee JS, Lee SY, Min JK, Shetty A, Park SH, Kim SJ, Cho ML. Small heterodimer partner-interacting leucine zipper protein suppresses pain and cartilage destruction in an osteoarthritis model by modulating the AMPK/STAT3 signaling pathway. Arthritis Res Ther 2024; 26:199. [PMID: 39533324 PMCID: PMC11555939 DOI: 10.1186/s13075-024-03417-3] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/15/2024] [Accepted: 10/16/2024] [Indexed: 11/16/2024] Open
Abstract
OBJECTIVE Osteoarthritis (OA) is a degenerative joint disease caused by the breakdown of joint cartilage and adjacent bone. Joint injury, being overweight, differences in leg length, high levels of joint stress, abnormal joint or limb development, and inherited factors have been implicated in the etiology of OA. In addition to physical damage to the joint, a role for inflammatory processes has been identified as well. Small heterodimer partner-interacting leucine zipper protein (SMILE) regulates transcription and many cellular functions. Among the proteins activated by SMILE is the peroxisome proliferator-activated receptor (PPAR) γ, which mediates the activities of CD4 + T helper cells, including Th1, Th2, and Th17, as well as Treg cells. PPAR-γ binds to STAT3 to inhibit its transcription, thereby suppressing the expression of the NF-κB pathway, and in turn, the expression of the inflammatory cytokines interferon (IFN), interleukin (IL)-1β, IL-6, and tumor necrosis factor (TNF)-α, which are sub-signals of STAT3 and NF-κB. METHODS OA was induced in control C57BL/6 mice and in C57BL/6-derived SMILE-overexpressing transgenic (SMILE Tg) mice. The protein expression levels in the joint and spleen tissues were analyzed by immunohistochemistry and immunofluorescence images. In addition, flow cytometry was performed for detecting changes of the changes of immune cells. RESULTS Less cartilage damage and significantly reduced levels of OA biomarkers (MMP13, TIMP3 and MCP-1) were observed in SMILE Tg mice. Immunohistochemistry performed to identify the signaling pathway involved in the link between SMILE expression and OA revealed decreased levels of IL-1β, IL-6, TNF-α, and phosphorylated AMPK in synovial tissues as well as a significant decrease in phosphorylated STAT3 in both cartilage and synovium. Changes in systemic immune cells were investigated via flow cytometry to analyze splenocytes isolated from control and SMILE Tg mice. SMILE Tg mice had elevated proportions of CD4 + IL-4 + cells (Th2) and CD4 + CD25 + Foxp3 + cells (Treg) and a notable decrease in CD4 + IL-17 + cells (Th17). CONCLUSION Our results show that overexpressed SMILE attenuates the symptoms of OA, by increasing AMPK signaling and decreasing STAT3, thus reducing the levels of inflammatory immune cells.
Collapse
Affiliation(s)
- Jeonghyeon Moon
- Departments of Immunobiology and Neurology, Yale School of Medicine, New Haven, CT, USA
| | - Keun-Hyung Cho
- Lab of Translational ImmunoMedicine (LaTIM), Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - JooYeon Jhun
- Lab of Translational ImmunoMedicine (LaTIM), Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - JeongWon Choi
- Lab of Translational ImmunoMedicine (LaTIM), Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Hyun-Sik Na
- Lab of Translational ImmunoMedicine (LaTIM), Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Jeong Su Lee
- Lab of Translational ImmunoMedicine (LaTIM), Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Seung Yoon Lee
- Lab of Translational ImmunoMedicine (LaTIM), Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea
| | - Jun-Ki Min
- Department of Internal Medicine, and the Clinical Medicine Research Institute of Bucheon St. Mary's Hospital, The Catholic University of Korea, Bucheon si, Gyeonggi-do, Korea
| | - Anan Shetty
- Institute of Medical Sciences, Canterbury Christ Church University, Medway Campus, Chatham, Kent, UK
| | - Sung-Hwan Park
- Department of Internal Medicine, Division of Rheumatology, College of Medicine, Seoul St. Mary's Hospital, The Catholic University of Korea, 222, Banpo‑daero, Seocho‑gu, Seoul, 06591, Korea
| | - Seok Jung Kim
- Department of Orthopaedic Surgery, Uijeongbu St. Mary's Hospital, College of Medicine, The Catholic University of Korea, Cheonbo-ro, Uijeongbu-si, Gyeonggi-do, 271, Korea
| | - Mi-La Cho
- Lab of Translational ImmunoMedicine (LaTIM), Catholic Research Institute of Medical Science, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
- Department of Pathology, College of Medicine, The Catholic University of Korea, Seoul, 06591, Korea.
| |
Collapse
|
7
|
Amodeo G, Magni G, Galimberti G, Riboldi B, Franchi S, Sacerdote P, Ceruti S. Neuroinflammation in osteoarthritis: From pain to mood disorders. Biochem Pharmacol 2024; 228:116182. [PMID: 38556026 DOI: 10.1016/j.bcp.2024.116182] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2024] [Revised: 03/19/2024] [Accepted: 03/28/2024] [Indexed: 04/02/2024]
Abstract
Osteoarthritis (OA) is the most common form of musculoskeletal disease, and its prevalence is increasing due to the aging of the population. Chronic pain is the most burdensome symptom of OA that significantly lowers patients' quality of life, also due to its frequent association with emotional comorbidities, such as anxiety and depression. In recent years, both chronic pain and mood alterations have been linked to the development of neuroinflammation in the peripheral nervous system, spinal cord and supraspinal brain areas. Thus, mechanisms at the basis of the development of the neuroinflammatory process may indicate promising targets for novel treatment for pain and affective comorbidities that accompany OA. In order to assess the key role of neuroinflammation in the maintenance of chronic pain and its potential involvement in development of psychiatric components, the monoiodoacetate (MIA) model of OA in rodents has been used and validated. In the present commentary article, we aim to summarize up-to-date results achieved in this experimental model of OA, focusing on glia activation and cytokine production in the sciatic nerve, dorsal root ganglia (DRGs), spinal cord and brain areas. The association of a neuroinflammatory state with the development of pain and anxiety- and depression-like behaviors are discussed. Results suggest that cells and molecules involved in neuroinflammation may represent novel targets for innovative pharmacological treatments of OA pain and mood comorbidities.
Collapse
Affiliation(s)
- Giada Amodeo
- Laboratory of Pain Therapy and Neuroimmunology, Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Via Balzaretti, 9 -20133 Milan (IT), Italy
| | - Giulia Magni
- Laboratory of Pain Therapy and Neuroimmunology, Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Via Balzaretti, 9 -20133 Milan (IT), Italy
| | - Giulia Galimberti
- Laboratory of Pain Therapy and Neuroimmunology, Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Via Balzaretti, 9 -20133 Milan (IT), Italy
| | - Benedetta Riboldi
- Laboratory of Pain Therapy and Neuroimmunology, Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Via Balzaretti, 9 -20133 Milan (IT), Italy
| | - Silvia Franchi
- Laboratory of Pain Therapy and Neuroimmunology, Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Via Balzaretti, 9 -20133 Milan (IT), Italy
| | - Paola Sacerdote
- Laboratory of Pain Therapy and Neuroimmunology, Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Via Balzaretti, 9 -20133 Milan (IT), Italy
| | - Stefania Ceruti
- Laboratory of Pain Therapy and Neuroimmunology, Department of Pharmacological and Biomolecular Sciences "Rodolfo Paoletti", Università degli Studi di Milano, Via Balzaretti, 9 -20133 Milan (IT), Italy.
| |
Collapse
|
8
|
Angrisani N, von der Ahe C, Willumeit-Römer R, Windhagen H, Scheper V, Schwarze M, Wiese B, Helmholz H, Reifenrath J. Treatment of osteoarthritis by implantation of Mg- and WE43-cylinders - A preclinical study on bone and cartilage changes and their influence on pain sensation in rabbits. Bioact Mater 2024; 40:366-377. [PMID: 38978802 PMCID: PMC11228885 DOI: 10.1016/j.bioactmat.2024.06.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2023] [Revised: 04/20/2024] [Accepted: 06/01/2024] [Indexed: 07/10/2024] Open
Abstract
With its main features of cartilage degeneration, subchondral bone sclerosis and osteophyte formation, osteoarthritis represents a multifactorial disease with no effective treatment options. As biomechanical shift in the trabecular network may be a driver for further cartilage degeneration, bone enhancement could possibly delay OA progression. Magnesium is known to be osteoconductive and already showed positive effects in OA models. We aimed to use magnesium cylinders to enhance subchondral bone quality, condition of cartilage and pain sensation compared to sole drilling in vivo. After eight weeks of implantation in rabbits, significant increase in subchondral bone volume and trabecular thickness with constant bone mineral density was found indicating favored biomechanics. As representative for pain, a higher number of CD271+ vessels were present in control samples without magnesium. However, this result could not be confirmed by sensitive, objective lameness evaluation using a pressure sensing mat and no positive effect could be shown on either cartilage degeneration evaluated by OARSI score nor the presence of regenerative cells in CD271-stained samples. The presented results show a relevant impact of implanted magnesium on key structures in OA pain with missing clinical relevance regarding pain. Further studies with shifted focus should examine additional structures as joint capsule or osteophytes.
Collapse
Affiliation(s)
- Nina Angrisani
- Hannover Medical School, Clinic for Orthopaedic Surgery, Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Lower Saxony, Germany
| | - Christin von der Ahe
- Hannover Medical School, Clinic for Orthopaedic Surgery, Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Lower Saxony, Germany
| | | | - Henning Windhagen
- Hannover Medical School, Clinic for Orthopaedic Surgery, Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Lower Saxony, Germany
| | - Verena Scheper
- Hannover Medical School, Department of Otolaryngology, Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Lower Saxony, Germany
| | - Michael Schwarze
- Hannover Medical School, Clinic for Orthopaedic Surgery, Laboratory for Biomechanics and Biomaterials, Hannover, Lower Saxony, Germany
| | - Björn Wiese
- Helmholtz-Zentrum Hereon, Institute of Metallic Biomaterials, Geesthacht, Germany
| | - Heike Helmholz
- Helmholtz-Zentrum Hereon, Institute of Metallic Biomaterials, Geesthacht, Germany
| | - Janin Reifenrath
- Hannover Medical School, Clinic for Orthopaedic Surgery, Lower Saxony Centre for Biomedical Engineering, Implant Research and Development (NIFE), Hannover, Lower Saxony, Germany
| |
Collapse
|
9
|
Brochard S, Boumédiene K, Mercier J, Agin V, Conrozier T, Baugé C. A single intraarticular injection of a tranexamic acid-modified hyaluronic acid (HA/TXA) alleviates pain and reduces OA development in a murine model of monosodium iodoacetate-induced osteoarthritis. Front Pharmacol 2024; 15:1456495. [PMID: 39323636 PMCID: PMC11422076 DOI: 10.3389/fphar.2024.1456495] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/28/2024] [Accepted: 08/26/2024] [Indexed: 09/27/2024] Open
Abstract
Rationale Tranexamic acid (TXA) is a strong and specific plasminogen activator inhibitor with inhibitory effects on the matrix metalloproteases involved in the pathophysiology of osteoarthritis (OA) through targeting of the fibrinolysis pathway. In this study, we evaluated the analgesic and chondroprotective effects of a HA-tranexamic acid (HA/TXA) conjugate, compared to HA alone and placebo, in an animal model of knee OA. Methods Knee OA was induced in 15 C57 b l/6J mice by IA injection of 0.75 mg of Monosodium IodoAcetate (MIA). At day 28, the mice received 1 IA injection of 10 µL of saline (control-group), or of HA or of HA/TXA. Tactile sensitivity was assessed using von Frey filaments. Stimulations started at 1 g and increased until a response was obtained (up to 4 g). A response to the stimulus was counted if the animal withdrew its paw. If the animal responded to the 1 g stimulation, stimulation was reduced until the lack of response was observed (up to 0.2 g). At day 56, mice were euthanized for knee histological assessment. Cartilage degradation was assessed using the OARSI score. Statistical analysis was performed on GraphPad Prism 8.0.2 software. Kruskal-Wallis or Mann-Whitney tests were performed as appropriate. Results Just before treatment administration, no intergroup difference in paw withdrawal threshold was observed. Throughout the experiment animals given saline and HA had a lower paw withdrawal threshold than those treated with HA/TXA (p < 0.01). In the control group OARSI score was 5.5 ± 0.6. In HA and HA + TXA treated mice the OARSI score was 3.2 ± 0.8 and 3.1 ± 0.5 (p < 0.01) showing that both treatments were able to reduce OA progression. Conclusion In this animal model of MIA induced KOA, a single IA injection of a HA/TXA conjugate resulted in a greater efficacy on pain than both saline and HA. HA and HA/TXA exhibited chondroprotective effects compared to placebo.
Collapse
Affiliation(s)
| | | | | | - Véronique Agin
- INSERM U1237, Physiopathology and Imaging of Neurological Disorders, Université de Caen Normandie, Caen, France
| | - Thierry Conrozier
- Department of Rheumatology, Hôpital Nord Franche-Comté, Belfort, France
| | - Catherine Baugé
- UR7451 Bioconnect, Université de Caen Normandie, Caen, France
| |
Collapse
|
10
|
Ventura L, do Espírito-Santo RF, Keaser M, Zhang Y, Ro JY, Da Silva JT. Green Light Exposure Reduces Primary Hyperalgesia and Proinflammatory Cytokines in a Rodent Model of Knee Osteoarthritis: Shedding Light on Sex Differences. Biomedicines 2024; 12:2005. [PMID: 39335519 PMCID: PMC11429231 DOI: 10.3390/biomedicines12092005] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/23/2024] [Revised: 08/20/2024] [Accepted: 08/26/2024] [Indexed: 09/30/2024] Open
Abstract
Knee osteoarthritis (OA) often causes chronic pain that disproportionately affects females. Proinflammatory cytokines TNF-α, IL-1β, and IL-6 are key effectors of OA pathological changes. Green light shows potential as an alternative intervention for various pain conditions. However, no studies have investigated green light's analgesic effects in both sexes in chronic knee OA. We induced unilateral knee OA with intra-articular injection of monoiodoacetate (MIA) in male and female Sprague-Dawley rats. Two days post-injection, the rats were exposed to green-light-emitting diodes (GLED) or ambient room light eight hours daily for 24 days. Knee mechanical sensitivity was assessed using a small animal algometer. Blood serum concentrations of TNF-α, IL-1β, IL-6, and IL-10 were quantified at baseline and 23 days post-injection. MIA injection decreased the knee mechanical thresholds of the male and female rats. GLED exposure attenuated mechanical hypersensitivity in both sexes compared to the controls; however, GLED-induced analgesia occurred sooner and with greater magnitude in males than in females. In both sexes, the analgesic effects of green light lasted 5 days after the final GLED session. Finally, GLED exposure reversed the elevation of serum proinflammatory cytokines. These findings suggest that GLED exposure reduces primary hyperalgesia in OA, potentially by lowering proinflammatory cytokines, and indicate sex differences in GLED-induced analgesia.
Collapse
Affiliation(s)
- Laura Ventura
- Center to Advance Chronic Pain Research, Department of Neural and Pain Sciences, University of Maryland Baltimore School of Dentistry, Baltimore, MD 21201, USA
- Program in Neuroscience, University of Maryland Baltimore School of Medicine, Baltimore, MD 21201, USA
| | - Renan F do Espírito-Santo
- Center to Advance Chronic Pain Research, Department of Neural and Pain Sciences, University of Maryland Baltimore School of Dentistry, Baltimore, MD 21201, USA
| | - Michael Keaser
- Center to Advance Chronic Pain Research, Department of Neural and Pain Sciences, University of Maryland Baltimore School of Dentistry, Baltimore, MD 21201, USA
| | - Youping Zhang
- Center to Advance Chronic Pain Research, Department of Neural and Pain Sciences, University of Maryland Baltimore School of Dentistry, Baltimore, MD 21201, USA
| | - Jin Y Ro
- Center to Advance Chronic Pain Research, Department of Neural and Pain Sciences, University of Maryland Baltimore School of Dentistry, Baltimore, MD 21201, USA
| | - Joyce T Da Silva
- Center to Advance Chronic Pain Research, Department of Neural and Pain Sciences, University of Maryland Baltimore School of Dentistry, Baltimore, MD 21201, USA
- Program in Neuroscience, University of Maryland Baltimore School of Medicine, Baltimore, MD 21201, USA
| |
Collapse
|
11
|
Mao H, Feng Y, Feng J, Yusufu Y, Sun M, Yang L, Jiang Q. Quercetin-3-O-β-D-glucuronide attenuates osteoarthritis by inhibiting cartilage extracellular matrix degradation and inflammation. J Orthop Translat 2024; 45:236-246. [PMID: 38601200 PMCID: PMC11004501 DOI: 10.1016/j.jot.2024.01.007] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 10/25/2023] [Revised: 01/21/2024] [Accepted: 01/31/2024] [Indexed: 04/12/2024] Open
Abstract
Objective Osteoarthritis (OA) is a chronic degenerative joint disease characterized by cartilage damage. In order to find a safer and more effective drug to treat OA, we investigated the role of quercetin-3-O-β-D-glucuronide (Q3GA) in OA. Methods We used qRT-PCR and western blots to detect the effects of Q3GA on extracellular matrix (ECM) and inflammation related genes and proteins in interleukin-1β (IL-1β) induced chondrocytes. We determined the effect of Q3GA on the NF-κB pathway using western blots and immunofluorescence. Moreover, the effect of Q3GA on the Nrf2 pathway was evaluated through molecular docking, western blots, and immunofluorescence experiments and further validated by transfection with Nrf2 siRNA. Subsequently, we established a rat model of OA and injected Q3GA into the joint cavity for treatment. After 5 weeks of Q3GA administration, samples were obtained for micro-computed tomography scanning and histopathological staining to determine the effects of Q3GA on OA rats. Results We found that Q3GA reduced the degradation of ECM and the expression of inflammatory related proteins and genes in primary chondrocytes of rats induced by IL-1β, as well as the expression of nitric oxide (NO) and reactive oxygen species (ROS). It inhibited the activation of the NF-κB pathway by increasing the expression of Nrf2 in the nucleus. In addition, Q3GA inhibited cartilage degradation in OA rats and promoted cartilage repair. Conclusion Q3GA attenuates OA by inhibiting ECM degradation and inflammation via the Nrf2/NF-κB axis. The translational potential of this article The results of our study demonstrate the promising potential of Q3GA as a candidate drug for the treatment of OA and reveal its key mechanisms.
Collapse
Affiliation(s)
- Haijun Mao
- Department of Orthopedic Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, China
| | - Yanwei Feng
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Juan Feng
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Yalikun Yusufu
- Department of Orthopedic Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, China
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, China
| | - Minghui Sun
- Department of Orthopedic Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, China
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, China
| | - Lei Yang
- Jiangsu Key Laboratory of Bioactive Natural Product Research and State Key Laboratory of Natural Medicines, School of Traditional Chinese Pharmacy, China Pharmaceutical University, Nanjing, 210009, China
| | - Qing Jiang
- Department of Orthopedic Surgery, Nanjing Drum Tower Hospital Clinical College of Nanjing Medical University, Nanjing, 210008, China
- Division of Sports Medicine and Adult Reconstructive Surgery, Department of Orthopedic Surgery, Nanjing Drum Tower Hospital, The Affiliated Hospital of Medical School, Nanjing University, Nanjing, 210008, China
- State Key Laboratory of Pharmaceutical Biotechnology, Nanjing University, Nanjing, 210008, China
- Branch of National Clinical Research Center for Orthopedics, Sports Medicine and Rehabilitation, China
| |
Collapse
|
12
|
Ma K, Pham T, Wang J, O-Sullivan I, DiCamillo A, Du S, Mwale F, Farooqui Z, Votta-Velis G, Bruce B, van Wijnen AJ, Liu Y, Im HJ. Nanoparticle-based inhibition of vascular endothelial growth factor receptors alleviates osteoarthritis pain and cartilage damage. SCIENCE ADVANCES 2024; 10:eadi5501. [PMID: 38354243 PMCID: PMC10866538 DOI: 10.1126/sciadv.adi5501] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2023] [Accepted: 01/12/2024] [Indexed: 02/16/2024]
Abstract
Osteoarthritis (OA) is characterized by cartilage damage, inflammation, and pain. Vascular endothelial growth factor receptors (VEGFRs) have been associated with OA severity, suggesting that inhibitors targeting these receptors alleviate pain (via VEGFR1) or cartilage degeneration (via VEGFR2). We have developed a nanoparticle-based formulation of pazopanib (Votrient), an FDA-approved anticancer drug that targets both VEGFR1 and VEGFR2 (Nano-PAZII). We demonstrate that a single intraarticular injection of Nano-PAZII can effectively reduce joint pain for a prolonged time without substantial side effects in two different preclinical OA rodent models involving either surgical (upon partial medial meniscectomy) or nonsurgical induction (with monoiodoacetate). The injection of Nano-PAZII blocks VEGFR1 and relieves OA pain by suppressing sensory neuronal ingrowth into the knee synovium and neuronal plasticity in the dorsal root ganglia and spinal cord. Simultaneously, the inhibition of VEGFR2 reduces cartilage degeneration. These findings provide a mechanism-based disease-modifying drug strategy that addresses both pain symptoms and cartilage loss in OA.
Collapse
Affiliation(s)
- Kaige Ma
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Tiep Pham
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, IL 60608, USA
| | - Jun Wang
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - InSug O-Sullivan
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Amy DiCamillo
- Melior Discovery Inc., 869 Springdale Drive 500, Exton, PA 19341, USA
| | - Shiyu Du
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, IL 60608, USA
| | - Fackson Mwale
- Orthopaedic Research Laboratory, Lady Davis Institute for Medical Research, SMBD-Jewish General Hospital, McGill University, Montreal, Canada
| | - Zeba Farooqui
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
| | - Gina Votta-Velis
- Department of Anesthesiology, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Benjamin Bruce
- Jesse Brown Veterans Affairs Medical Center (JBVAMC) at Chicago, IL 60612, USA
| | - Andre J. van Wijnen
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
- Department of Biochemistry, University of Vermont, Burlington, VT 05405, USA
| | - Ying Liu
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
- Department of Chemical Engineering, University of Illinois at Chicago, Chicago, IL 60608, USA
- Department of Pharmaceutical Sciences, University of Illinois at Chicago, Chicago, IL 60612, USA
| | - Hee-Jeong Im
- Department of Biomedical Engineering, University of Illinois at Chicago, Chicago, IL 60607, USA
- Jesse Brown Veterans Affairs Medical Center (JBVAMC) at Chicago, IL 60612, USA
| |
Collapse
|
13
|
Zhang YZ, Wei ZJ, Yu SN, Wang XY, Wang Y, Wu CA, Jiang X. Dihydrotanshinone I protects human chondrocytes and alleviates damage from spontaneous osteoarthritis in a guinea pig model. Sci Rep 2023; 13:21355. [PMID: 38049518 PMCID: PMC10696037 DOI: 10.1038/s41598-023-48902-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/20/2023] [Accepted: 11/30/2023] [Indexed: 12/06/2023] Open
Abstract
Osteoarthritis (OA) is the most common degenerative joint disease. Currently, no satisfactory pharmacological treatment exists for OA. The potential anti-inflammatory properties of Dihydrotanshinone I (DHT) have been reported, but its effects on OA are unclear. In this study, we assess the impact of DHT on the viability of human chondrocytes in vitro. We then use a guinea pig model to investigate the effects of DHT on knee osteoarthritis progression. Twelve-week-old Dunkin Hartley guinea pigs spontaneously developing OA were intraperitoneally injected with different doses of DHT for eight weeks. Micro-CT analysis was performed on the subchondral bone in the knee, and histological assessment of the knee joint was done using stained sections, the ratio of hyaline to calcified cartilage, and Mankin scores. DHT successfully restored IL-1β-induced decreases in cell viability in human primary chondrocytes. In the guinea pig model, intraperitoneal injections of DHT ameliorated age-induced OA, effectively reduced the expression level of two cartilage metabolism-related genes (ADAMTS4 and MMP13) and decreased the inflammatory biomarker IL-6 in the serum of guinea pigs developing spontaneous osteoarthritis. These findings demonstrate DHT's protective effects on chondrocytes and suggest that it alleviates cartilage degradation and proteoglycan loss in OA.
Collapse
Affiliation(s)
- Yan-Zhuo Zhang
- National Center for Orthopaedics, Department of Molecular Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, People's Republic of China
| | - Zhen-Jie Wei
- National Center for Orthopaedics, Department of Molecular Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, People's Republic of China
| | - Shu-Nan Yu
- National Center for Orthopaedics, Department of Molecular Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, People's Republic of China
| | - Xin-Yu Wang
- National Center for Orthopaedics, Department of Molecular Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, People's Republic of China
| | - Ying Wang
- National Center for Orthopaedics, Department of Molecular Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, People's Republic of China
| | - Cheng-Ai Wu
- National Center for Orthopaedics, Department of Molecular Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, People's Republic of China
| | - Xu Jiang
- National Center for OrthopaedicsDepartment of Orthopaedics, Beijing Research Institute of Traumatology and Orthopaedics, Beijing Jishuitan Hospital, Capital Medical University, Beijing, 100035, People's Republic of China.
| |
Collapse
|
14
|
Choi WW, Kim SH, Kim JH, Kim K, Kim SJ, Kim M, Kim HS, Lee H, Lee JY, Yong SY. Preclinical Study of Dual-Wavelength Light-Emitting Diode Therapy in an Osteoarthritis Rat Model. Ann Rehabil Med 2023; 47:483-492. [PMID: 38053342 PMCID: PMC10767218 DOI: 10.5535/arm.23138] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/22/2023] [Revised: 10/26/2023] [Accepted: 11/01/2023] [Indexed: 12/07/2023] Open
Abstract
OBJECTIVE To evaluate the efficacy of light-emitting diode (LED) and their dual-wavelengths as a treatment strategy for osteoarthritis. METHODS We induced osteoarthritis in male Sprague-Dawley rats by intra-articular injection of sodium iodoacetate into the right rear knee joint. The animals with lesions were divided into an untreated group and an LED-treated group (n=7 each). In the LED-treated group, the lesioned knee was irradiated with lasers (850 and 940 nm) and dose (3.15 J/cm2) for 20 minutes per session, twice a week for 4 weeks. Knee joint tissues were stained and scanned using an in vivo micro-computed tomography (CT) scanner. Serum interleukin (IL)-6 and IL-18 levels were determined using enzyme-linked immuno-sorbent assay. Several functional tests (lines crossed, rotational movement, rearing, and latency to remain rotating rod) were performed 24 hours before LED treatment and at 7, 14, 21, and 28 days after treatment. RESULTS LED-treated rats showed improved locomotor function and suppressed matrix-degrading cytokines. Micro-CT images indicated that LED therapy had a preserving effect on cartilage and cortical bone. CONCLUSION LED treatment using wavelengths of 850 and 940 nm resulted in significant functional, anatomical, and histologic improvements without adverse events in a rat model. Further research is required to determine the optimal wavelength, duration, and combination method, which will maximize treatment effectiveness.
Collapse
Affiliation(s)
- Won Woo Choi
- Department of Rehabilitation Medicine, Wonju Severance Christian Hospital, Wonju, Korea
| | - Sung Hoon Kim
- Department of Rehabilitation Medicine, Wonju Severance Christian Hospital, Wonju, Korea
- Yonsei Institute of Sports Science and Exercise Medicine, Wonju, Korea
| | - Ji Hyun Kim
- Department of Rehabilitation Medicine, Wonju Severance Christian Hospital, Wonju, Korea
- Yonsei Institute of Sports Science and Exercise Medicine, Wonju, Korea
| | - Kyungmin Kim
- Department of Rehabilitation Medicine, Wonju Severance Christian Hospital, Wonju, Korea
| | - Sun Jung Kim
- Department of Rehabilitation Medicine, Wonju Severance Christian Hospital, Wonju, Korea
| | - Minwoo Kim
- Department of Rehabilitation Medicine, Wonju Severance Christian Hospital, Wonju, Korea
| | - Han-Sung Kim
- Department of Biomedical Engineering, Yonsei University, Wonju, Korea
| | - Hana Lee
- Department of Biomedical Engineering, Yonsei University, Wonju, Korea
| | - Ji Yong Lee
- Research Institute of Hyperbaric Medicine and Science, Yonsei University Wonju College of Medicine, Wonju, Korea
| | - Sang Yeol Yong
- Department of Rehabilitation Medicine, Wonju Severance Christian Hospital, Wonju, Korea
- Yonsei Institute of Sports Science and Exercise Medicine, Wonju, Korea
| |
Collapse
|
15
|
Sun J, Wang XH, Song FH, Li DY, Gao SJ, Zhang LQ, Wu JY, Liu DQ, Wang LW, Zhou YQ, Mei W. Inhibition of Brd4 alleviates osteoarthritis pain via suppression of neuroinflammation and activation of Nrf2-mediated antioxidant signalling. Br J Pharmacol 2023; 180:3194-3214. [PMID: 37485568 DOI: 10.1111/bph.16195] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/17/2022] [Revised: 05/02/2023] [Accepted: 05/03/2023] [Indexed: 07/25/2023] Open
Abstract
BACKGROUND AND PURPOSE Osteoarthritis (OA) pain remains a major clinical problem. It is urgent to identify novel therapeutic approaches for OA pain states. Bromodomain and extra-terminal (BET) protein inhibitors have robust anti-inflammatory effects in several pain models. However, the underlying mechanisms of these inhibitors in OA pain have not been determined. We, therefore, investigated the effects and the underlying mechanism(s) of BET inhibition on pain-related behaviours in a rat model of OA. EXPERIMENTAL APPROACH The OA model was established by intra-articular injection of monosodium iodoacetate (MIA) in rat knees. Pain behaviours were assessed in rats by hindlimb weight-bearing asymmetry, mechanical allodynia and thermal hyperalgesia. Possible mechanisms underlying BET inhibition were explored in the MIA-induced OA pain model in the spinal cord and dorsal root ganglia (DRG). KEY RESULTS Inhibiting bromodomain-containing protein 4 (Brd4) with either JQ1 or MS417, or using AAV2/9-shRNA-Brd4-EGFP-mediated knockdown of Brd4 genes, significantly attenuated MIA-induced pain behaviours. Brd4 inhibition suppressed NF-κB and NF-κB-mediated inflammatory cytokines in both the spinal cord and DRG in rats with MIA-induced OA pain. Brd4 inhibition also attenuated the oxidative stress and promoted nuclear factor erythroid-2-related factor 2 (Nrf2)-dependent antioxidant genes in both the spinal cord and DRG in our odel of MIA-induced OA pain. CONCLUSIONS AND IMPLICATIONS In conclusion, Brd4 inhibition alleviated MIA-induced OA pain in rats, via suppression of neuroinflammation and activation of Nrf2-mediated antioxidant signalling. Although our model does not perfectly represent how OA develops in humans, inhibition of Brd4 may provide novel insights into possible treatments for OA pain.
Collapse
Affiliation(s)
- Jia Sun
- Department of Anesthesiology, Xuzhou Central Hospital, Xuzhou, China
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Xing-He Wang
- Department of Anesthesiology, Xuzhou Central Hospital, Xuzhou, China
| | - Fan-He Song
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dan-Yang Li
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Shao-Jie Gao
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Long-Qing Zhang
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Jia-Yi Wu
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Dai-Qiang Liu
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Li-Wei Wang
- Department of Anesthesiology, Xuzhou Central Hospital, Xuzhou, China
| | - Ya-Qun Zhou
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| | - Wei Mei
- Department of Anesthesiology, Hubei Key Laboratory of Geriatric Anesthesia and Perioperative Brain Health, and Wuhan Clinical Research Center for Geriatric Anesthesia, Tongji Hospital, Tongji Medical College, Huazhong University of Science and Technology, Wuhan, China
| |
Collapse
|
16
|
Geraghty T, Obeidat AM, Ishihara S, Wood MJ, Li J, Lopes EBP, Scanzello CR, Griffin TM, Malfait AM, Miller RE. Age-Associated Changes in Knee Osteoarthritis, Pain-Related Behaviors, and Dorsal Root Ganglia Immunophenotyping of Male and Female Mice. Arthritis Rheumatol 2023; 75:1770-1780. [PMID: 37096632 PMCID: PMC10543384 DOI: 10.1002/art.42530] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/12/2022] [Revised: 02/07/2023] [Accepted: 03/28/2023] [Indexed: 04/26/2023]
Abstract
OBJECTIVE Osteoarthritis (OA) is a leading cause of chronic pain, yet OA pain management remains poor. Age is the strongest predictor of OA development, and mechanisms driving OA pain are unclear. We undertook this study to characterize age-associated changes in knee OA, pain-related behaviors, and dorsal root ganglion (DRG) molecular phenotypes in mice of both sexes. METHODS Male or female C57BL/6 mice 6 or 20 months of age were evaluated for histopathologic knee OA, pain-related behaviors, and L3-L5 DRG immune characterization via flow cytometry. DRG gene expression in older mice and humans was also examined. RESULTS Male mice at 20 months of age had worse cartilage degeneration than 6-month-old mice. Older female mouse knees showed increased cartilage degeneration but to a lesser degree than those of male mice. Older mice of both sexes had worse mechanical allodynia, knee hyperalgesia, and grip strength compared to younger mice. For both sexes, DRGs from older mice showed decreased CD45+ cells and a significant increase in F4/80+ macrophages and CD11c+ dendritic cells. Older male mouse DRGs showed increased expression of Ccl2 and Ccl5, and older female mouse DRGs showed increased Cxcr4 and Ccl3 expression compared to 6-month-old mouse DRGs, among other differentially expressed genes. Human DRG analysis from 6 individuals >80 years of age revealed elevated CCL2 in men compared to women, whereas CCL3 was higher in DRGs from women. CONCLUSION We found that aging in male and female mice is accompanied by mild knee OA, mechanical sensitization, and changes to immune cell populations in the DRG, suggesting novel avenues for development of OA therapies.
Collapse
Affiliation(s)
- Terese Geraghty
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, USA
| | - Alia M. Obeidat
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, USA
| | - Shingo Ishihara
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, USA
| | - Matthew J. Wood
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, USA
| | - Jun Li
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, USA
| | | | - Carla R. Scanzello
- Department of Medicine, Division of Rheumatology, University of Pennsylvania, Philadelphia, PA, USA
- Translational Musculoskeletal Research Center, Corp. Michael J. Crescenz VA Medical Center, Philadelphia, PA
| | - Timothy M. Griffin
- Oklahoma Medical Research Foundation, Oklahoma City, OK, USA
- OKC Veterans Affairs Medical Center, Oklahoma City, OK, USA
| | - Anne-Marie Malfait
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, USA
| | - Rachel E. Miller
- Department of Internal Medicine, Division of Rheumatology, Rush University Medical Center, Chicago, IL, USA
| |
Collapse
|
17
|
Deng C, Chen Y, Zhao X, Yu L, Xiao Y, Li H, Zhang Y, Ai K, Zhou D, Bai X, Gong T, Wei J, Zeng C, Lei G. Apoptotic Neutrophil Membrane-Camouflaged Liposomes for Dually Targeting Synovial Macrophages and Fibroblasts to Attenuate Osteoarthritis. ACS APPLIED MATERIALS & INTERFACES 2023; 15:39064-39080. [PMID: 37523857 DOI: 10.1021/acsami.3c05861] [Citation(s) in RCA: 2] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 08/02/2023]
Abstract
No current pharmacological approach is capable of simultaneously inhibiting the symptomatology and structural progression of osteoarthritis. M1 macrophages and activated synovial fibroblasts (SFs) mutually contribute to the propagation of joint pain and cartilage destruction in osteoarthritis. Here, we report the engineering of an apoptotic neutrophil membrane-camouflaged liposome (termed "NM@Lip") for precise delivery of triamcinolone acetonide (TA) by dually targeting M1 macrophages and activated SFs in osteoarthritic joints. NM@Lip has a high cellular uptake in M1 macrophages and activated SFs. Furthermore, TA-loaded NM@Lip (TA-NM@Lip) effectively repolarizes M1 macrophages to the M2 phenotype and transforms pathological SFs to the deactivated phenotype by inhibiting the PI3K/Akt pathway. NM@Lip retains in the joint for up to 28 days and selectively distributes into M1 macrophages and activated SFs in synovium with low distribution in cartilage. TA-NM@Lip decreases the levels of pro-inflammatory cytokines, chemokines, and cartilage-degrading enzymes in osteoarthritic joints. In a rodent model of osteoarthritis-related pain, a single intra-articular TA-NM@Lip injection attenuates synovitis effectively and achieves complete pain relief with long-lasting effects. In a rodent model of osteoarthritis-related joint degeneration, repeated intra-articular TA-NM@Lip injections induce no obvious cartilage damage and effectively attenuate cartilage degeneration. Taken together, TA-NM@Lip represents a promising nanotherapeutic approach for osteoarthritis therapy.
Collapse
Affiliation(s)
- Caifeng Deng
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha 410008, China
| | - Yuxiao Chen
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha 410008, China
| | - Xuan Zhao
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha 410008, China
| | - Liukang Yu
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha 410008, China
| | - Yongbing Xiao
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha 410008, China
| | - Hui Li
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha 410008, China
| | - Yuqing Zhang
- Division of Rheumatology, Allergy, and Immunology, Department of Medicine, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, United States
- The Mongan Institute, Massachusetts General Hospital, Harvard Medical School, Boston, Massachusetts 02114, United States
| | - Kelong Ai
- Xiangya School of Pharmaceutical Sciences, Central South University, Changsha 410078, China
| | - Dongfang Zhou
- NMPA Key Laboratory for Research and Evaluation of Drug Metabolism & Guangdong Provincial Key Laboratory of New Drug Screening, School of Pharmaceutical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Xiaochun Bai
- Guangdong Provincial Key Laboratory of Bone and Joint Degeneration Diseases, Department of Cell Biology, School of Basic Medical Sciences, Southern Medical University, Guangzhou 510515, China
| | - Tao Gong
- Key Laboratory of Drug-Targeting and Drug Delivery System of the Education Ministry, Sichuan Engineering Laboratory for Plant-Sourced Drug and Sichuan Research Center for Drug Precision Industrial Technology, West China School of Pharmacy, Sichuan University, Chengdu 610064, China
| | - Jie Wei
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha 410008, China
- Health Management Center, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Chao Zeng
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| | - Guanghua Lei
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha 410008, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Changsha 410008, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha 410008, China
| |
Collapse
|
18
|
Jin H, Yang Y, Lei G, Zeng C, He K, Wang Y, Deng C, Wei J, Li X, Li H. Pain Intensity and Trajectory Following Intra-Articular Injection of Mono-Iodoacetate in Experimental Osteoarthritis: A Meta-Analysis of In Vivo Studies. Cartilage 2023; 14:86-93. [PMID: 36628407 PMCID: PMC10076896 DOI: 10.1177/19476035221144748] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023] Open
Abstract
OBJECTIVE Although most frequently used in experimental osteoarthritis (OA) pain induction, intra-articular mono-iodoacetate (MIA) injection lacks concluded references for dose selection and timing of intervention. Herein, we aimed to compare the pain intensity of rats induced by different doses of MIA and explored the trajectory of pain. DESIGN PubMed, Embase, and Web of Science were searched up to June 2021 for literatures involving MIA experiments investigating OA pain. Pain intensity was measured based on weightbearing distribution (WBD) and paw withdrawal thresholds (PWT), and the pain trajectory was constructed by evaluating pain intensity at a series of time points after MIA injection. A conventional meta-analysis was conducted. RESULTS A total of 140 studies were included. Compared with saline, MIA injections caused significantly higher pain intensity for WBD and PWT. Dose-response relationships between different doses of MIA and pain intensity were observed (P-for-trend<0.05). A pronounced increase in pain occurred from day 0 to day 7, but the uptrend ceased between day 7 and day 14, after which the pain intensity continued to rise and reached the maximum by day 28. CONCLUSIONS Pain intensity after intra-articular MIA injection increased in a dose-dependent manner and the pain trajectory manifested a specific pattern consistent with the pathological mechanisms of MIA-induced pain, providing possible clues for proper dose selection and timing of specific OA pain interventions.
Collapse
Affiliation(s)
- Hongyu Jin
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Yuanheng Yang
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Department of Plastic and Cosmetic Surgery, Xiangya Hospital, Central South University, Changsha, China
| | - Guanghua Lei
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Chao Zeng
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
- National Clinical Research Center for Geriatric Disorders, Xiangya Hospital, Central South University, Changsha, China
| | - Ke He
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
| | - Yilun Wang
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| | - Caifeng Deng
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
| | - Jie Wei
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
- Health Management Center, Xiangya Hospital, Central South University, Changsha, China
| | - Xiaoxiao Li
- Hunan Key Laboratory of Joint Degeneration and Injury, Xiangya Hospital, Central South University, Changsha, China
| | - Hui Li
- Department of Orthopaedics, Xiangya Hospital, Central South University, Changsha, China
| |
Collapse
|
19
|
Chung MK, Wang S, Alshanqiti I, Hu J, Ro JY. The degeneration-pain relationship in the temporomandibular joint: Current understandings and rodent models. FRONTIERS IN PAIN RESEARCH 2023; 4:1038808. [PMID: 36846071 PMCID: PMC9947567 DOI: 10.3389/fpain.2023.1038808] [Citation(s) in RCA: 5] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2022] [Accepted: 01/18/2023] [Indexed: 02/11/2023] Open
Abstract
Temporomandibular disorders (TMD) represent a group of musculoskeletal conditions involving the temporomandibular joints (TMJ), the masticatory muscles and associated structures. Painful TMD are highly prevalent and conditions afflict 4% of US adults annually. TMD include heterogenous musculoskeletal pain conditions, such as myalgia, arthralgia, and myofascial pain. A subpopulations of TMD patients show structural changes in TMJ, including disc displacement or degenerative joint diseases (DJD). DJD is a slowly progressing, degenerative disease of the TMJ characterized by cartilage degradation and subchondral bone remodeling. Patients with DJD often develop pain (TMJ osteoarthritis; TMJ OA), but do not always have pain (TMJ osteoarthrosis). Therefore, pain symptoms are not always associated with altered TMJ structures, which suggests that a causal relationship between TMJ degeneration and pain is unclear. Multiple animal models have been developed for determining altered joint structure and pain phenotypes in response to various TMJ injuries. Rodent models of TMJOA and pain include injections to induce inflammation or cartilage destruction, sustained opening of the oral cavity, surgical resection of the articular disc, transgenic approaches to knockout or overexpress key genes, and an integrative approach with superimposed emotional stress or comorbidities. In rodents, TMJ pain and degeneration occur during partially overlapping time periods in these models, which suggests that common biological factors may mediate TMJ pain and degeneration over different time courses. While substances such as intra-articular pro-inflammatory cytokines commonly cause pain and joint degeneration, it remains unclear whether pain or nociceptive activities are causally associated with structural degeneration of TMJ and whether structural degeneration of TMJ is necessary for producing persistent pain. A thorough understanding of the determining factors of pain-structure relationships of TMJ during the onset, progression, and chronification by adopting novel approaches and models should improve the ability to simultaneously treat TMJ pain and TMJ degeneration.
Collapse
Affiliation(s)
- Man-Kyo Chung
- Department of Neural and Pain Sciences, School of Dentistry, Program in Neuroscience, Center to Advance Chronic Pain Research, University of Maryland Baltimore, Baltimore, MD, United States
| | | | | | | | | |
Collapse
|
20
|
Riewruja K, Makarczyk M, Alexander PG, Gao Q, Goodman SB, Bunnell BA, Gold MS, Lin H. Experimental models to study osteoarthritis pain and develop therapeutics. OSTEOARTHRITIS AND CARTILAGE OPEN 2022; 4:100306. [PMID: 36474784 PMCID: PMC9718172 DOI: 10.1016/j.ocarto.2022.100306] [Citation(s) in RCA: 2] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/06/2022] [Revised: 07/25/2022] [Accepted: 08/09/2022] [Indexed: 10/15/2022] Open
Abstract
Pain is the predominant symptom of osteoarthritis (OA) that drives patients to seek medical care. Currently, there are no pharmacological treatments that can reverse or halt the progression of OA. Safe and efficacious medications for long-term management of OA pain are also unavailable. Understanding the mechanisms behind OA pain generation at onset and over time is critical for developing effective treatments. In this narrative review, we first summarize our current knowledge on the innervation of the knee joint, and then discuss the molecular mechanism(s) currently thought to underlie OA pain. In particular, we focus on the contribution of each joint component to the generation of pain. Next, the current experimental models for studying OA pain are summarized, and the methods to assess pain in rodents are presented. The potential application of emerging microphysiological systems in OA pain research is especially highlighted. Lastly, we discuss the current challenge in standardizing models and the selection of appropriate systems to address specific questions.
Collapse
Affiliation(s)
- Kanyakorn Riewruja
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Osteoarthritis and Musculoskeleton Research Unit, Faculty of Medicine, Chulalongkorn University, King Chulalongkorn Memorial Hospital, Thai Red Cross Society, Bangkok, 10330, Thailand
| | - Meagan Makarczyk
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, USA
| | - Peter G. Alexander
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Qi Gao
- Department of Orthopaedic Surgery, Stanford, CA, USA
| | | | - Bruce A. Bunnell
- Department of Microbiology, Immunology, and Genetics, University of North Texas Health Science Center, Fort Worth, TX, USA
| | - Michael S. Gold
- Department of Neurobiology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
| | - Hang Lin
- Department of Orthopaedic Surgery, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Bioengineering, University of Pittsburgh Swanson School of Engineering, Pittsburgh, PA, USA
| |
Collapse
|
21
|
Cuffaro D, Ciccone L, Rossello A, Nuti E, Santamaria S. Targeting Aggrecanases for Osteoarthritis Therapy: From Zinc Chelation to Exosite Inhibition. J Med Chem 2022; 65:13505-13532. [PMID: 36250680 PMCID: PMC9620172 DOI: 10.1021/acs.jmedchem.2c01177] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/22/2022] [Indexed: 11/30/2022]
Abstract
Osteoarthritis (OA) is the most common degenerative joint disease. In 1999, two members of the A Disintegrin and Metalloproteinase with Thrombospondin Motifs (ADAMTS) family of metalloproteinases, ADAMTS4 and ADAMTS5, or aggrecanases, were identified as the enzymes responsible for aggrecan degradation in cartilage. The first aggrecanase inhibitors targeted the active site by chelation of the catalytic zinc ion. Due to the generally disappointing performance of zinc-chelating inhibitors in preclinical and clinical studies, inhibition strategies tried to move away from the active-site zinc in order to improve selectivity. Exosite inhibitors bind to proteoglycan-binding residues present on the aggrecanase ancillary domains (called exosites). While exosite inhibitors are generally more selective than zinc-chelating inhibitors, they are still far from fulfilling their potential, partly due to a lack of structural and functional data on aggrecanase exosites. Filling this gap will inform the design of novel potent, selective aggrecanase inhibitors.
Collapse
Affiliation(s)
- Doretta Cuffaro
- Department
of Pharmacy, University of Pisa, via Bonanno 6, 56126 Pisa, Italy
| | - Lidia Ciccone
- Department
of Pharmacy, University of Pisa, via Bonanno 6, 56126 Pisa, Italy
| | - Armando Rossello
- Department
of Pharmacy, University of Pisa, via Bonanno 6, 56126 Pisa, Italy
| | - Elisa Nuti
- Department
of Pharmacy, University of Pisa, via Bonanno 6, 56126 Pisa, Italy
| | - Salvatore Santamaria
- Department
of Immunology and Inflammation, Imperial
College London, Du Cane Road, London W12
0NN, U.K.
| |
Collapse
|
22
|
Induction of Accelerated Aging in a Mouse Model. Cells 2022; 11:cells11091418. [PMID: 35563724 PMCID: PMC9102583 DOI: 10.3390/cells11091418] [Citation(s) in RCA: 13] [Impact Index Per Article: 6.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/26/2022] [Revised: 04/11/2022] [Accepted: 04/20/2022] [Indexed: 12/12/2022] Open
Abstract
With the global increase of the elderly population, the improvement of the treatment for various aging-related diseases and the extension of a healthy lifespan have become some of the most important current medical issues. In order to understand the developmental mechanisms of aging and aging-related disorders, animal models are essential to conduct relevant studies. Among them, mice have become one of the most prevalently used model animals for aging-related studies due to their high similarity to humans in terms of genetic background and physiological structure, as well as their short lifespan and ease of reproduction. This review will discuss some of the common and emerging mouse models of accelerated aging and related chronic diseases in recent years, with the aim of serving as a reference for future application in fundamental and translational research.
Collapse
|